51
|
Nishizuka SS, Mills GB. New era of integrated cancer biomarker discovery using reverse-phase protein arrays. Drug Metab Pharmacokinet 2016; 31:35-45. [DOI: 10.1016/j.dmpk.2015.11.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/26/2015] [Accepted: 11/29/2015] [Indexed: 12/11/2022]
|
52
|
Wagner T, Kiweler N, Wolff K, Knauer SK, Brandl A, Hemmerich P, Dannenberg JH, Heinzel T, Schneider G, Krämer OH. Sumoylation of HDAC2 promotes NF-κB-dependent gene expression. Oncotarget 2016; 6:7123-35. [PMID: 25704882 PMCID: PMC4466673 DOI: 10.18632/oncotarget.3344] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 01/04/2015] [Indexed: 01/15/2023] Open
Abstract
The transcription factor nuclear factor-κB (NF-κB) is crucial for the maintenance of homeostasis. It is incompletely understood how nuclear NF-κB and the crosstalk of NF-κB with other transcription factors are controlled. Here, we demonstrate that the epigenetic regulator histone deacetylase 2 (HDAC2) activates NF-κB in transformed and primary cells. This function depends on both, the catalytic activity and an intact HDAC2 sumoylation motif. Several mechanisms account for the induction of NF-κB through HDAC2. The expression of wild-type HDAC2 can increase the nuclear presence of NF-κB. In addition, the ribosomal S6 kinase 1 (RSK1) and the tumor suppressor p53 contribute to the regulation of NF-κB by HDAC2. Moreover, TP53 mRNA expression is positively regulated by wild-type HDAC2 but not by sumoylation-deficient HDAC2. Thus, sumoylation of HDAC2 integrates NF-κB signaling involving p53 and RSK1. Since HDAC2-dependent NF-κB activity protects colon cancer cells from genotoxic stress, our data also suggest that high HDAC2 levels, which are frequently found in tumors, are linked to chemoresistance. Accordingly, inhibitors of NF-κB and of the NF-κB/p53-regulated anti-apoptotic protein survivin significantly sensitize colon carcinoma cells expressing wild-type HDAC2 to apoptosis induced by the genotoxin doxorubicin. Hence, the HDAC2-dependent signaling node we describe here may offer an interesting therapeutic option.
Collapse
Affiliation(s)
- Tobias Wagner
- Centre for Molecular Biomedicine, Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University of Jena, Jena, Germany
| | - Nicole Kiweler
- Department of Toxicology, University Medical Center, Mainz, Germany
| | - Katharina Wolff
- Centre for Molecular Biomedicine, Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University of Jena, Jena, Germany
| | - Shirley K Knauer
- Centre for Medical Biotechnology, Molecular Biology II, University of Duisburg-Essen, Essen, Germany
| | - André Brandl
- Centre for Molecular Biomedicine, Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University of Jena, Jena, Germany
| | - Peter Hemmerich
- Leibniz-Institute for Age Research, Fritz-Lipmann-Institute, Jena, Germany
| | - Jan-Hermen Dannenberg
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Thorsten Heinzel
- Centre for Molecular Biomedicine, Institute of Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University of Jena, Jena, Germany
| | - Günter Schneider
- Klinikum rechts der Isar, II. Medizinische Klinik, Technische Universität München, München, Germany
| | - Oliver H Krämer
- Department of Toxicology, University Medical Center, Mainz, Germany
| |
Collapse
|
53
|
Hu Y, Ge W, Wang X, Sutendra G, Zhao K, Dedeić Z, Slee EA, Baer C, Lu X. Caspase cleavage of iASPP potentiates its ability to inhibit p53 and NF-κB. Oncotarget 2015; 6:42478-90. [PMID: 26646590 PMCID: PMC4767446 DOI: 10.18632/oncotarget.6478] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/25/2015] [Indexed: 12/14/2022] Open
Abstract
An intriguing biological question relating to cell signaling is how the inflammatory mediator NF-kB and the tumour suppressor protein p53 can be induced by similar triggers, like DNA damage or infection, yet have seemingly opposing or sometimes cooperative biological functions. For example, the NF-κB subunit RelA/p65 has been shown to inhibit apoptosis, whereas p53 induces apoptosis. One potential explanation may be their co-regulation by common cellular factors: inhibitor of Apoptosis Stimulating p53 Protein (iASPP) is one such common regulator of both RelA/p65 and p53. Here we show that iASPP is a novel substrate of caspases in response to apoptotic stimuli. Caspase cleaves the N-terminal region of iASPP at SSLD294 resulting in a prominent 80kDa fragment of iASPP. This caspase cleavage site is conserved in various species from zebrafish to Homo sapiens. The 80kDa fragment of iASPP translocates from the cytoplasm to the nucleus via the RaDAR nuclear import pathway, independent of p53. The 80kDa iASPP fragment can bind and inhibit p53 or RelA/p65 more efficiently than full-length iASPP. Overall, these data reveal a potential novel regulation of p53 and RelA/p65 activities in response to apoptotic stimuli.
Collapse
Affiliation(s)
- Ying Hu
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Wenjie Ge
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xingwen Wang
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Gopinath Sutendra
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Kunming Zhao
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zinaida Dedeić
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Elizabeth A. Slee
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Caroline Baer
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Xin Lu
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
54
|
Jmjd5 functions as a regulator of p53 signaling during mouse embryogenesis. Cell Tissue Res 2015; 363:723-33. [DOI: 10.1007/s00441-015-2276-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/08/2015] [Indexed: 01/21/2023]
|
55
|
Lin Y, Mallen-St Clair J, Luo J, Sharma S, Dubinett S, St John M. p53 modulates NF-κB mediated epithelial-to-mesenchymal transition in head and neck squamous cell carcinoma. Oral Oncol 2015; 51:921-8. [PMID: 26306422 DOI: 10.1016/j.oraloncology.2015.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/15/2015] [Accepted: 07/17/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVES To investigate the role of p53 in NF-κB mediated epithelial-to-mesenchymal (EMT) in head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS We utilized HNSCC and normal oral epithelial cell lines as our model system. We used a lentiviral shRNA system to silence the expression of p65 and p53 in these cell lines. Mutant and wild-type (WT) p53 background genotypes were analyzed. The expression of epithelial and mesenchymal markers was determined using western blotting and quantitative PCR assays. Cell morphology, growth, and invasion were determined using a 3-dimensional spheroid culture and anchorage independent growth (AIG) assays. RESULTS In HNSCC cells with mutant p53 we found that silencing p65 expression promoted EMT. In contrast, in the context of WT p53, ectopic p65 over-expression promoted EMT. Ablation of WT p53 in normal oral epithelial cells blocked EMT induced by p65 over-expression. We demonstrate that AIG and apoptosis induced by NF-κB activation is regulated by p53. CONCLUSION Our data demonstrates that p53 mutational status is critical in determining the outcome of NF-κB activation in HNSCC. In the presence of WT p53, excess p65 signal can promote EMT. Conversely, ablation of p65 in the context of mutant p53 drives EMT. These results demonstrate that p53 mutational status alters the outcome of NF-κB signaling. These results, though preliminary, demonstrate the critical role of p53 mutational status in determining the outcome of NF-κB signaling and suggest that monitoring p53 status may inform the utility of NF-κB inhibitor treatment in HNSCC.
Collapse
Affiliation(s)
- Yuan Lin
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - Jon Mallen-St Clair
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - Jie Luo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - Sherven Sharma
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Veterans' Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States
| | - Steven Dubinett
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Veterans' Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States
| | - Maie St John
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States.
| |
Collapse
|
56
|
TLR2∆22 (-196-174) significantly increases the risk of breast cancer in females carrying proline allele at codon 72 of TP53 gene: a case-control study from four ethnic groups of North Eastern region of India. Tumour Biol 2015; 36:9995-10002. [PMID: 26188904 DOI: 10.1007/s13277-015-3795-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/10/2015] [Indexed: 12/13/2022] Open
Abstract
Breast cancer (BC) is the second most common cancer in women. In the North Eastern Region (NER) of India, BC is emerging as an important concern as evidenced by the data available from population and hospital-based cancer registries. Studies on genetic susceptibility to BC are important to understand the increase in the incidence of BC in NER. The present case control study was conducted to investigate the association between tumour suppressor gene TP53 codon 72 polymorphism and innate immune pathway gene TLR2∆22 (-196-174) polymorphism with BC in females of NER of India for the identification of novel biomarker of BC. Four hundred sixty-two histopathologically confirmed BC cases from four states of NER of India, and 770 healthy controls were included by organizing community surveys from the neighbourhood of cases. In our study, no significant association between TP53 codon 72 polymorphisms and the risk of BC was found. However, our study has shown that TP53 codon 72 polymorphism is an important effect modifier. In the present study it was found that females carrying 22 base-pair deletion in the promoter region of their TLR2 gene had two times (AOR= 2.18, 95 % CI 1.13-4.21, p=0.019 in dominant model; AOR= 2.17, 95 % CI 1.09-4.34, p=0.027 in co-dominant model) increased risk of BC whwn they also carry proline allele at codon 72 of their TP53 gene.
Collapse
|
57
|
Association of TP53 gene polymorphisms with susceptibility of bladder cancer in Bangladeshi population. Tumour Biol 2015; 36:6369-74. [DOI: 10.1007/s13277-015-3324-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 03/12/2015] [Indexed: 01/23/2023] Open
|
58
|
McGraw KL, Zhang LM, Rollison DE, Basiorka AA, Fulp W, Rawal B, Jerez A, Billingsley DL, Lin HY, Kurtin SE, Yoder S, Zhang Y, Guinta K, Mallo M, Solé F, Calasanz MJ, Cervera J, Such E, González T, Nevill TJ, Haferlach T, Smith AE, Kulasekararaj A, Mufti G, Karsan A, Maciejewski JP, Sokol L, Epling-Burnette PK, Wei S, List AF. The relationship of TP53 R72P polymorphism to disease outcome and TP53 mutation in myelodysplastic syndromes. Blood Cancer J 2015; 5:e291. [PMID: 25768405 PMCID: PMC4382654 DOI: 10.1038/bcj.2015.11] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/13/2015] [Indexed: 01/22/2023] Open
Abstract
Nonsynonymous TP53 exon 4 single-nucleotide polymorphism (SNP), R72P, is linked to cancer and mutagen susceptibility. R72P associations with specific cancer risk, particularly hematological malignancies, have been conflicting. Myelodysplastic syndrome (MDS) with chromosome 5q deletion is characterized by erythroid hypoplasia arising from lineage-specific p53 accumulation resulting from ribosomal insufficiency. We hypothesized that apoptotically diminished R72P C-allele may influence predisposition to del(5q) MDS. Bone marrow and blood DNA was sequenced from 705 MDS cases (333 del(5q), 372 non-del(5q)) and 157 controls. Genotype distribution did not significantly differ between del(5q) cases (12.6% CC, 38.1% CG, 49.2% GG), non-del(5q) cases (9.7% CC, 44.6% CG, 45.7% GG) and controls (7.6% CC, 37.6% CG, 54.8% GG) (P=0.13). Allele frequency did not differ between non-del(5q) and del(5q) cases (P=0.91) but trended towards increased C-allele frequency comparing non-del(5q) (P=0.08) and del(5q) (P=0.10) cases with controls. Median lenalidomide response duration increased proportionate to C-allele dosage in del(5q) patients (2.2 (CC), 1.3 (CG) and 0.89 years (GG)). Furthermore, C-allele homozygosity in del(5q) was associated with prolonged overall and progression-free survival and non-terminal interstitial deletions that excluded 5q34, whereas G-allele homozygozity was associated with inferior outcome and terminal deletions involving 5q34 (P=0.05). These findings comprise the largest MDS R72P SNP analysis.
Collapse
Affiliation(s)
- K L McGraw
- Hematology Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - L M Zhang
- Molecular Genomics Core Lab, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - D E Rollison
- Cancer Epidemiology, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - A A Basiorka
- 1] Hematology Department, H Lee Moffitt Cancer Center, Tampa, FL, USA [2] Cancer Biology PhD Program, University of South Florida, Tampa, FL, USA
| | - W Fulp
- Biostatistics and Bioinformatics Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - B Rawal
- Mayo Clinic, Biostatistics-Division of Health Sciences Research, Jacksonville, FL, USA
| | - A Jerez
- Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, USA
| | | | - H-Y Lin
- Biostatistics and Bioinformatics Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | - S Yoder
- Molecular Genomics Core Lab, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Y Zhang
- Biostatistics and Bioinformatics Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - K Guinta
- Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, USA
| | - M Mallo
- Institut de Recerca Contra la Leucèmia Josep Carreras (IJC) Badalona, Barcelona, Spain
| | - F Solé
- Institut de Recerca Contra la Leucèmia Josep Carreras (IJC) Badalona, Barcelona, Spain
| | - M J Calasanz
- Hematology Department, Hospital Universitario La Fe, Valencia, Spain
| | - J Cervera
- Hematology Department, Hospital Universitario La Fe, Valencia, Spain
| | - E Such
- Hematology Department, Hospital Universitario La Fe, Valencia, Spain
| | - T González
- Genomics Medicine Public Foundation, Hospital Clinico Universitario, Santiago de Compostela, Spain
| | - T J Nevill
- British Columbia Cancer Agency, Vancouver, BC, Canada
| | | | - A E Smith
- King's College London, King's College Hospital, London, UK
| | | | - G Mufti
- King's College London, King's College Hospital, London, UK
| | - A Karsan
- British Columbia Cancer Agency, Vancouver, BC, Canada
| | - J P Maciejewski
- Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, USA
| | - L Sokol
- Hematology Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | - S Wei
- Immunology Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - A F List
- Hematology Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
59
|
Barabutis N, Dimitropoulou C, Birmpas C, Joshi A, Thangjam G, Catravas JD. p53 protects against LPS-induced lung endothelial barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 2015; 308:L776-87. [PMID: 25713322 DOI: 10.1152/ajplung.00334.2014] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/09/2015] [Indexed: 12/16/2022] Open
Abstract
New therapies toward heart and blood vessel disorders may emerge from the development of Hsp90 inhibitors. Several independent studies suggest potent anti-inflammatory activities of those agents in human tissues. The molecular mechanisms responsible for their protective effects in the vasculature remain unclear. The present study demonstrates that the transcription factor p53, an Hsp90 client protein, is crucial for the maintenance of vascular integrity, protects again LPS-induced endothelial barrier dysfunction, and is involved in the mediation of the anti-inflammatory activity of Hsp90 inhibitors in lung tissues. p53 silencing by siRNA decreased transendothelial resistance (a measure of endothelial barrier function). A similar effect was induced by the p53 inhibitor pifithrin, which also potentiated the LPS-induced hyperpermeability in human lung microvascular endothelial cells (HLMVEC). On the other hand, p53 induction by nutlin suppressed the LPS-induced vascular barrier dysfunction. LPS decreased p53 expression in lung tissues and that effect was blocked by pretreatment with Hsp90 inhibitors both in vivo and in vitro. Furthermore, the Hsp90 inhibitor 17-allyl-amino-demethoxy-geldanamycin suppressed the LPS-induced overexpression of the p53 negative regulator MDMX as well as p53 and MDM2 (another p53 negative regulator) phosphorylation in HLMVEC. Both negative p53 regulators were downregulated by LPS in vivo. Chemically induced p53 overexpression resulted in the suppression of LPS-induced RhoA activation and MLC2 phosphorylation, whereas p53 suppression caused the opposite effects. These observations reveal new mechanisms for the anti-inflammatory actions of Hsp90 inhibitors, i.e., the induction of the transcription factor p53, which in turn can orchestrate robust vascular anti-inflammatory responses both in vivo and in vitro.
Collapse
Affiliation(s)
| | | | | | - Atul Joshi
- Frank Reidy Research Center for Bioelectrics, Norfolk, Virginia; and
| | - Gagan Thangjam
- Frank Reidy Research Center for Bioelectrics, Norfolk, Virginia; and
| | - John D Catravas
- Frank Reidy Research Center for Bioelectrics, Norfolk, Virginia; and School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, Virginia
| |
Collapse
|
60
|
NF-κB regulates caspase-4 expression and sensitizes neuroblastoma cells to Fas-induced apoptosis. PLoS One 2015; 10:e0117953. [PMID: 25695505 PMCID: PMC4335045 DOI: 10.1371/journal.pone.0117953] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/06/2015] [Indexed: 12/01/2022] Open
Abstract
Found in neurons and neuroblastoma cells, Fas-induced apoptosis and accompanied activation of NF-κB signaling were thought to be associated with neurodegenerative diseases. However, the detailed functions of NF-κB activation in Fas killing and the effect of NF-κB activation on its downstream events remain unclear. Here, we demonstrated that agonistic Fas antibody induces cell death in a dose-dependent way and NF-κB signaling is activated as well, in neuroblastoma cells SH-EP1. Unexpectedly, NF-κB activation was shown to be pro-apoptotic, as suggested by the reduction of Fas-induced cell death with either a dominant negative form of IκBα (DN-IκBα) or an IκB kinase-specific inhibitor. To our interest, when analyzing downstream events of NF-κB signaling, we found that DN-IκBα only suppressed the expression of caspase-4, but not other caspases. Vice versa, enhancement of NF-κB activity by p65 (RelA) overexpression increased the expression of caspase-4 at both mRNA and protein levels. More directly, results from dual luciferase reporter assay demonstrated the regulation of caspase-4 promoter activity by NF-κB. When caspase-4 activity was blocked by its dominant negative (DN) form, Fas-induced cell death was substantially reduced. Consistently, the cleavage of PARP and caspase-3 induced by Fas was also reduced. In contrast, the cleavage of caspase-8 remained unaffected in caspase-4 DN cells, although caspase-8 inhibitor could rescue Fas-induced cell death. Collectively, these data suggest that caspase-4 activity is required for Fas-induced cell apoptosis and caspase-4 may act upstream of PARP and caspase-3 and downstream of caspase-8. Overall, we demonstrate that NF-κB can mediate Fas-induced apoptosis through caspase-4 protease, indicating that caspase-4 is a new mediator of NF-κB pro-apoptotic pathway in neuroblastoma cells.
Collapse
|
61
|
Abstract
The tumor-suppressor protein p53 belongs to a family of proteins that play pivotal roles in multiple cellular functions including cell proliferation, cell death, genome stability, and regulation of inflammation. Neuroinflammation is a common feature of central nervous system (CNS) pathology, and microglia are the specialized resident population of CNS myeloid cells that initiate innate immune responses. Microglia maintain CNS homeostasis through pathogen containment, phagocytosis of debris, and initiation of tissue-repair cascades. However, an unregulated pro-inflammatory response can lead to tissue injury and dysfunction in both acute and chronic inflammatory states. Therefore, regulation of the molecular signals that control the induction, magnitude, and resolution of inflammation are necessary for optimal CNS health. We and others have described a novel mechanism by which p53 transcriptional activity modulates microglia behaviors in vitro and in vivo. Activation of p53 induces expression of microRNAs (miRNAs) that support microglia pro-inflammatory functions and suppress anti-inflammatory and tissue repair behaviors. In this review, we introduce the previously described roles of the p53 signaling network and discuss novel functions of p53 in the microglia-mediated inflammatory response in CNS health and disease. Ultimately, improved understanding of the molecular regulators modulated by p53 transcriptional activity in microglia will enhance the development of rational therapeutic strategies to harness the homeostatic and tissue repair functions of microglia.
Collapse
Affiliation(s)
- Macarena S. Aloi
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Wei Su
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Gwenn A. Garden
- Department of Pathology, University of Washington, Seattle, Washington, USA
- Department of Neurology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
62
|
Chand HS, Montano G, Huang X, Randell SH, Mebratu Y, Petersen H, Tesfaigzi Y. A genetic variant of p53 restricts the mucous secretory phenotype by regulating SPDEF and Bcl-2 expression. Nat Commun 2014; 5:5567. [PMID: 25429397 PMCID: PMC4247165 DOI: 10.1038/ncomms6567] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/13/2014] [Indexed: 02/07/2023] Open
Abstract
Despite implications for carcinogenesis and other chronic diseases, basic mechanisms of p53 and its variants in suppressing Bcl-2 levels are poorly understood. Bcl-2 sustains mucous cell metaplasia, whereas p53(-/-) mice display chronically increased mucous cells. Here we show that p53 decreases bcl-2 mRNA half-life by interacting with the 5' untranslated region (UTR). The p53-bcl-2 mRNA interaction is modified by the substitution of proline by arginine within the p53 proline-rich domain (PRD). Accordingly, more mucous cells are present in primary human airway cultures with p53(Arg) compared with p53(Pro). Also, the p53(Arg) compared with p53(Pro) displays higher affinity to and activates the promoter region of SAM-pointed domain-containing Ets-like factor (SPDEF), a driver of mucous differentiation. On two genetic backgrounds, mice with targeted replacement of prolines in p53 PRD show enhanced expression of SPDEF and Bcl-2 and mucous cell metaplasia. Together, these studies define the PRD of p53 as a determinant for chronic mucous hypersecretion.
Collapse
Affiliation(s)
- Hitendra S. Chand
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| | - Gilbert Montano
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| | - Xuesong Huang
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| | - Scott H. Randell
- Department of Cell and Molecular Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yohannes Mebratu
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| | - Hans Petersen
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| | - Yohannes Tesfaigzi
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| |
Collapse
|
63
|
Association between polymorphisms in tumor suppressor genes and oncogenes and risk of hepatocellular carcinoma: a case-control study in an HCC epidemic area within the Han Chinese population. Med Oncol 2014; 31:356. [PMID: 25412941 DOI: 10.1007/s12032-014-0356-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/12/2014] [Indexed: 02/08/2023]
Abstract
Data concerning the risk of hepatocellular carcinoma (HCC) and specific single nucleotide polymorphisms (SNPs) in an HBV-free population are currently limited. Therefore, we performed a case-control study to investigate the association between SNPs and the risk of HCC in individuals without chronic HBV infection. A total of 160 Han Chinese patients with HCC and an identical number of healthy controls were enrolled in this study. rs1042522, rs10814325, rs17401966, and rs2279744 genotypes were determined using matrix-associated laser desorption ionization-time of flight-mass spectrometry (MALDI-TOF-MS). CG and GG genotypes in rs1042522 and heterozygote and homozygote in rs2279744 were significantly associated with an elevated risk of HCC. Homozygous mutation of rs1081432 conferred a 2.68-fold risk of HCC (95% CI 1.35-5.34); however heterozygosity was not statistically significant. rs17401966 heterozygosity or homozygosity was not significantly associated with a increased risk of HCC. Several polymorphisms associated with a significantly increased risk of HCC were identified. These may serve as biomarkers in evaluating HCC risk in the general population.
Collapse
|
64
|
Panni S, Salvioli S, Santonico E, Langone F, Storino F, Altilia S, Franceschi C, Cesareni G, Castagnoli L. The adapter protein CD2AP binds to p53 protein in the cytoplasm and can discriminate its polymorphic variants P72R. J Biochem 2014; 157:101-11. [PMID: 25261582 DOI: 10.1093/jb/mvu059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Proline-rich motifs are widely distributed in eukaryotic proteomes and are usually involved in the assembly of functional complexes through interaction with specific binding modules. The tumour-suppressor p53 protein presents a proline-rich region that is crucial for regulating apoptosis by connecting the p53 with a complex protein network. In humans, a common polymorphism determines the identity of residue 72, either proline or arginine, and affects the features of the motifs present in the polyproline domain. The two isoforms have different biochemical properties and markedly influence cancer onset and progression. In this article, we analyse the binding of the p53 proline-rich region with a pool of selected polyproline binding domains (i.e. SH3 and WW), and we present the first demonstration that the purified SH3 domains of the CD2AP/Cin85 protein family are able to directly bind the p53 protein, and to discriminate between the two polymorphic variants P72R.
Collapse
Affiliation(s)
- Simona Panni
- Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy
| | - Stefano Salvioli
- Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy
| | - Elena Santonico
- Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy
| | - Francesca Langone
- Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy
| | - Francesca Storino
- Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy
| | - Serena Altilia
- Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy
| | - Claudio Franceschi
- Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy
| | - Gianni Cesareni
- Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy
| | - Luisa Castagnoli
- Department DiBEST, University of Calabria, Rende, 87036, Italy; DIMES, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy; CIG, Interdepartmental Center "Luigi Galvani", University of Bologna, Bologna 40126, Italy; Department of Biology, University of Rome Tor Vergata, Rome 00100, Italy; and Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, 00100, Italy
| |
Collapse
|
65
|
Kung CP, Khaku S, Jennis M, Zhou Y, Murphy ME. Identification of TRIML2, a novel p53 target, that enhances p53 SUMOylation and regulates the transactivation of proapoptotic genes. Mol Cancer Res 2014; 13:250-62. [PMID: 25256710 DOI: 10.1158/1541-7786.mcr-14-0385] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED The tumor-suppressor protein p53, encoded by TP53, inhibits tumorigenesis by inducing cell-cycle arrest, senescence, and apoptosis. Several genetic polymorphisms exist in TP53, including a proline to arginine variant at amino acid 72 (P72 and R72, respectively); this polymorphism alters p53 function. In general, the P72 variant shows increased ability to induce cell-cycle arrest, whereas the R72 variant possesses increased ability to induce apoptosis, relative to P72. At present, the underlying mechanisms for these functional differences are not fully understood. Toward elucidating the molecular basis for these differences, a gene-expression microarray analysis was conducted on normal human fibroblast cells that are homozygous for P72 and R72 variants, along with subclones of these lines that express a p53 short hairpin (shp53). Approximately three dozen genes were identified whose transactivation is affected by the codon 72 polymorphism. One of these is the tripartite-motif family-like 2 (TRIML2) gene, which is preferentially induced by the R72 variant. Importantly, the accumulated data indicate that TRIML2 interacts with p53, and facilitates the modification of p53 with SUMO2. TRIML2 also enhances the ability of p53 to transactivate a subset of proapoptotic target genes associated with prolonged oxidative stress, including PIDD, PIG3 (TP53I3), and PIG6 (PRODH). These data indicate that TRIML2 is part of a feed-forward loop that activates p53 in cells expressing the R72 variant, particularly after prolonged stress. IMPLICATIONS The defined actions of TRIML2, in part, explain the underlying molecular basis for increased apoptotic potential of the R72 variant of p53.
Collapse
Affiliation(s)
- Che-Pei Kung
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Sakina Khaku
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Matthew Jennis
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania. Drexel University College of Medicine, Program in Molecular Cell Biology and Genetics, Philadelphia, Pennsylvania
| | - Yan Zhou
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Maureen E Murphy
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania.
| |
Collapse
|
66
|
Iannetti A, Ledoux AC, Tudhope SJ, Sellier H, Zhao B, Mowla S, Moore A, Hummerich H, Gewurz BE, Cockell SJ, Jat PS, Willmore E, Perkins ND. Regulation of p53 and Rb links the alternative NF-κB pathway to EZH2 expression and cell senescence. PLoS Genet 2014; 10:e1004642. [PMID: 25255445 PMCID: PMC4177746 DOI: 10.1371/journal.pgen.1004642] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 07/28/2014] [Indexed: 11/18/2022] Open
Abstract
There are two major pathways leading to induction of NF-κB subunits. The classical (or canonical) pathway typically leads to the induction of RelA or c-Rel containing complexes, and involves the degradation of IκBα in a manner dependent on IκB kinase (IKK) β and the IKK regulatory subunit NEMO. The alternative (or non-canonical) pathway, involves the inducible processing of p100 to p52, leading to the induction of NF-κB2(p52)/RelB containing complexes, and is dependent on IKKα and NF-κB inducing kinase (NIK). Here we demonstrate that in primary human fibroblasts, the alternative NF-κB pathway subunits NF-κB2 and RelB have multiple, but distinct, effects on the expression of key regulators of the cell cycle, reactive oxygen species (ROS) generation and protein stability. Specifically, following siRNA knockdown, quantitative PCR, western blot analyses and chromatin immunoprecipitation (ChIP) show that NF-κB2 regulates the expression of CDK4 and CDK6, while RelB, through the regulation of genes such as PSMA5 and ANAPC1, regulates the stability of p21WAF1 and the tumour suppressor p53. These combine to regulate the activity of the retinoblastoma protein, Rb, leading to induction of polycomb protein EZH2 expression. Moreover, our ChIP analysis demonstrates that EZH2 is also a direct NF-κB target gene. Microarray analysis revealed that in fibroblasts, EZH2 antagonizes a subset of p53 target genes previously associated with the senescent cell phenotype, including DEK and RacGAP1. We show that this pathway provides the major route of crosstalk between the alternative NF-κB pathway and p53, a consequence of which is to suppress cell senescence. Importantly, we find that activation of NF-κB also induces EZH2 expression in CD40L stimulated cells from Chronic Lymphocytic Leukemia patients. We therefore propose that this pathway provides a mechanism through which microenvironment induced NF-κB can inhibit tumor suppressor function and promote tumorigenesis. Although the classical NF-κB pathway is frequently associated with the induction of cellular senescence and the senescence associated secretory phenotype (SASP), the role of the alternative NF-κB pathway, which is frequently activated in hematological malignancies as well as some solid tumors, has not been defined. We therefore investigated the role of the alternative NF-κB pathway in this process. Here we report that NF-κB2 and RelB, the effectors of the alternative NF-κB pathway, suppress senescence through inhibition of p53 activity. Using primary human fibroblasts, we demonstrate that this is accomplished through NF-κB2/RelB dependent control of a previously unknown pathway, incorporating regulation of CDK4 and 6 expression as well as regulators of p21WAF1 and p53 protein stability. Loss of NF-κB2/RelB results in suppression of retinoblastoma (Rb) tumour suppressor phosphorylation, which in turn leads to inhibition of EZH2 expression and de-repression of p53 activity. Interestingly, we find that CD40 ligand stimulation of cells from Chronic Lymphocytic Leukemia patients, which strongly induces the alternative NF-κB pathway, also induces EZH2 expression. We propose that the alternative NF-κB pathway can promote tumorigenesis through suppression of p53 dependent senescence, a process that may have relevance to cancer cells retaining wild type p53.
Collapse
Affiliation(s)
- Alessio Iannetti
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Adeline C. Ledoux
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Susan J. Tudhope
- Northern Institute for Cancer Research, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Hélène Sellier
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Bo Zhao
- Division of Infectious Disease, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Sophia Mowla
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Adam Moore
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Holger Hummerich
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Benjamin E. Gewurz
- Division of Infectious Disease, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Simon J. Cockell
- Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Parmjit S. Jat
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Elaine Willmore
- Northern Institute for Cancer Research, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Neil D. Perkins
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
- * E-mail:
| |
Collapse
|
67
|
Cooks T, Harris CC, Oren M. Caught in the cross fire: p53 in inflammation. Carcinogenesis 2014; 35:1680-90. [PMID: 24942866 PMCID: PMC4123652 DOI: 10.1093/carcin/bgu134] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 06/06/2014] [Accepted: 06/10/2014] [Indexed: 12/17/2022] Open
Abstract
The p53 transcription factor is a major tumor suppressor, whose diverse activities serve to ensure genome stability and inhibit neoplastic processes. In recent years, it is becoming increasingly clear that p53 also plays a broader role in maintaining cellular homeostasis, as well as contributing to tissue homeostasis in a non-cell-autonomous fashion. Chronic inflammation is a potential cancer-promoting condition, and as such is also within the radar of p53, which mounts a multifaceted attempt to prevent the escalation of chronic tissue imbalance into neoplasia. Recent understanding of the p53 pathway and other family members reveals a broad interaction with inflammatory elements such as reactive oxygen and nitrogen species, cytokines, infectious agents and major immune-regulatory pathways like nuclear factor-kappaB. This complex cross talk is highly dependent on p53 status, as different p53 isoforms and p53 mutants can mediate different responses and even promote chronic inflammation and associated cancer, acting in the tumor cells as well as in the stromal and immune compartments.
Collapse
Affiliation(s)
- Tomer Cooks
- Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD 20892-4258, USA and Molecular Cell Biology, Weizmann Institute for Science, Rehovot 76100, Israel
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD 20892-4258, USA and Molecular Cell Biology, Weizmann Institute for Science, Rehovot 76100, Israel
| | - Moshe Oren
- Molecular Cell Biology, Weizmann Institute for Science, Rehovot 76100, Israel
| |
Collapse
|
68
|
Lowe JM, Menendez D, Bushel PR, Shatz M, Kirk EL, Troester MA, Garantziotis S, Fessler MB, Resnick MA. p53 and NF-κB coregulate proinflammatory gene responses in human macrophages. Cancer Res 2014; 74:2182-92. [PMID: 24737129 DOI: 10.1158/0008-5472.can-13-1070] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Macrophages are sentinel immune cells that survey the tissue microenvironment, releasing cytokines in response to both exogenous insults and endogenous events such as tumorigenesis. Macrophages mediate tumor surveillance and therapy-induced tumor regression; however, tumor-associated macrophages (TAM) and their products may also promote tumor progression. Whereas NF-κB is prominent in macrophage-initiated inflammatory responses, little is known about the role of p53 in macrophage responses to environmental challenge, including chemotherapy or in TAMs. Here, we report that NF-κB and p53, which generally have opposing effects in cancer cells, coregulate induction of proinflammatory genes in primary human monocytes and macrophages. Using Nutlin-3 as a tool, we demonstrate that p53 and NF-κB rapidly and highly induce interleukin (IL)-6 by binding to its promoter. Transcriptome analysis revealed global p53/NF-κB co-regulation of immune response genes, including several chemokines, which effectively induced human neutrophil migration. In addition, we show that p53, activated by tumor cell paracrine factors, induces high basal levels of macrophage IL-6 in a TAM model system [tumor-conditioned macrophages (TCM)]. Compared with normal macrophages, TCMs exhibited higher p53 levels, enhanced p53 binding to the IL-6 promoter, and reduced IL-6 levels upon p53 inhibition. Taken together, we describe a mechanism by which human macrophages integrate signals through p53 and NF-κB to drive proinflammatory cytokine induction. Our results implicate a novel role for macrophage p53 in conditioning the tumor microenvironment and suggest a potential mechanism by which p53-activating chemotherapeutics, acting upon p53-sufficient macrophages and precursor monocytes, may indirectly impact tumors lacking functional p53.
Collapse
Affiliation(s)
- Julie M Lowe
- Authors' Affiliations: Laboratory of Molecular Genetics, Biostatistics Branch, Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park; and Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Weige CC, Birtwistle MR, Mallick H, Yi N, Berrong Z, Cloessner E, Duff K, Tidwell J, Clendenning M, Wilkerson B, Farrell C, Bunz F, Ji H, Shtutman M, Creek KE, Banister CE, Buckhaults PJ. Transcriptomes and shRNA suppressors in a TP53 allele-specific model of early-onset colon cancer in African Americans. Mol Cancer Res 2014; 12:1029-41. [PMID: 24743655 DOI: 10.1158/1541-7786.mcr-13-0286-t] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
UNLABELLED African Americans are disproportionately affected by early-onset, high-grade malignancies. A fraction of this cancer health disparity can be explained by genetic differences between individuals of African or European descent. Here the wild-type Pro/Pro genotype at the TP53Pro72Arg (P72R) polymorphism (SNP: rs1042522) is more frequent in African Americans with cancer than in African Americans without cancer (51% vs. 37%), and is associated with a significant increase in the rates of cancer diagnosis in African Americans. To test the hypothesis that Tp53 allele-specific gene expression may contribute to African American cancer disparities, TP53 hemizygous knockout variants were generated and characterized in the RKO colon carcinoma cell line, which is wild type for TP53 and heterozygous at the TP53Pro72Arg locus. Transcriptome profiling, using RNAseq, in response to the DNA-damaging agent etoposide revealed a large number of Tp53-regulated transcripts, but also a subset of transcripts that were TP53Pro72Arg allele specific. In addition, a shRNA-library suppressor screen for Tp53 allele-specific escape from Tp53-induced arrest was performed. Several novel RNAi suppressors of Tp53 were identified, one of which, PRDM1β (BLIMP-1), was confirmed to be an Arg-specific transcript. Prdm1β silences target genes by recruiting H3K9 trimethyl (H3K9me3) repressive chromatin marks, and is necessary for stem cell differentiation. These results reveal a novel model for African American cancer disparity, in which the TP53 codon 72 allele influences lifetime cancer risk by driving damaged cells to differentiation through an epigenetic mechanism involving gene silencing. IMPLICATIONS TP53 P72R polymorphism significantly contributes to increased African American cancer disparity.
Collapse
Affiliation(s)
| | - Marc R Birtwistle
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Himel Mallick
- Biostatistics, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Nengjun Yi
- Biostatistics, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Zuzana Berrong
- Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, Georgia
| | - Emily Cloessner
- Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, Georgia
| | - Keely Duff
- Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, Georgia
| | - Josephine Tidwell
- Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, Georgia
| | - Megan Clendenning
- Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, Georgia
| | - Brent Wilkerson
- Department of Otolaryngology-Head and Neck Surgery University of California, Davis, Sacramento, California
| | - Christopher Farrell
- Department of Pharmaceutical and Administrative Sciences School of Pharmacy, Presbyterian College, Clinton
| | - Fred Bunz
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hao Ji
- Department of Drug Discovery and Biomedical Sciences South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina; and
| | - Michael Shtutman
- Department of Drug Discovery and Biomedical Sciences South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina; and
| | - Kim E Creek
- Department of Drug Discovery and Biomedical Sciences South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina; and
| | - Carolyn E Banister
- Department of Drug Discovery and Biomedical Sciences South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina; and
| | | |
Collapse
|
70
|
Endo F, Nishizuka SS, Kume K, Ishida K, Katagiri H, Ishida K, Sato K, Iwaya T, Koeda K, Wakabayashi G. A compensatory role of NF-κB to p53 in response to 5-FU-based chemotherapy for gastric cancer cell lines. PLoS One 2014; 9:e90155. [PMID: 24587255 PMCID: PMC3937424 DOI: 10.1371/journal.pone.0090155] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 01/28/2014] [Indexed: 01/09/2023] Open
Abstract
Despite of remarkable improvement of postoperative 5-FU–based adjuvant chemotherapy, the relapse rate of gastric cancer patients who undergo curative resection followed by the adjuvant chemotherapy remains substantial. Therefore, it is important to identify prediction markers for the chemotherapeutic efficacy of 5-FU. We recently identified NF-κB as a candidate relapse prediction biomarker in gastric cancer. To evaluate the biological significance of NF-κB in the context of 5-FU–based chemotherapy, we analyzed the NF-κB-dependent biological response upon 5-FU treatment in gastric cancer cell lines. Seven genes induced by 5-FU treatment in an NF-κB-dependent manner were identified, five of which are known p53 targets. Knockdown of RELA, which encodes the p65 subunit of NF-κB, decreased both p53 and p53 target protein levels. In contrast, NF-κB was not affected by TP53 knockdown. We also demonstrated that cell lines bearing Pro/Pro homozygosity in codon72 of p53 exon4, which is important for NF-κB binding to p53, are more resistant to 5-FU than those with Arg/Arg homozygosity. We conclude that NF-κB plays an important role in the response to 5-FU treatment in gastric cancer cell lines, with a possible compensatory function of p53. These results suggest that NF-κB is a potential 5-FU-chemosensitivity prediction marker that may reflect 5-FU-induced stress-response pathways, including p53.
Collapse
Affiliation(s)
- Fumitaka Endo
- Molecular Therapeutics Laboratory, Iwate Medical University School of Medicine, Morioka, Japan
- Department of Surgery, Iwate Medical University School of Medicine, Morioka, Japan
| | - Satoshi S. Nishizuka
- Molecular Therapeutics Laboratory, Iwate Medical University School of Medicine, Morioka, Japan
- Department of Surgery, Iwate Medical University School of Medicine, Morioka, Japan
- MIAST (Medical Innovation by Advanced Science and Technology) project, Iwate Medical University, Morioka, Japan
- Institute for Biomedical Sciences, Iwate Medical University, Yahaba, Japan
- * E-mail:
| | - Kohei Kume
- Molecular Therapeutics Laboratory, Iwate Medical University School of Medicine, Morioka, Japan
- Department of Surgery, Iwate Medical University School of Medicine, Morioka, Japan
- MIAST (Medical Innovation by Advanced Science and Technology) project, Iwate Medical University, Morioka, Japan
- Institute for Biomedical Sciences, Iwate Medical University, Yahaba, Japan
| | - Kazushige Ishida
- Molecular Therapeutics Laboratory, Iwate Medical University School of Medicine, Morioka, Japan
- Department of Surgery, Iwate Medical University School of Medicine, Morioka, Japan
| | - Hirokatsu Katagiri
- Molecular Therapeutics Laboratory, Iwate Medical University School of Medicine, Morioka, Japan
- Department of Surgery, Iwate Medical University School of Medicine, Morioka, Japan
| | - Kaoru Ishida
- Molecular Therapeutics Laboratory, Iwate Medical University School of Medicine, Morioka, Japan
- Department of Surgery, Iwate Medical University School of Medicine, Morioka, Japan
| | - Kei Sato
- Molecular Therapeutics Laboratory, Iwate Medical University School of Medicine, Morioka, Japan
- Department of Surgery, Iwate Medical University School of Medicine, Morioka, Japan
| | - Takeshi Iwaya
- Molecular Therapeutics Laboratory, Iwate Medical University School of Medicine, Morioka, Japan
- Department of Surgery, Iwate Medical University School of Medicine, Morioka, Japan
| | - Keisuke Koeda
- Department of Surgery, Iwate Medical University School of Medicine, Morioka, Japan
| | - Go Wakabayashi
- Department of Surgery, Iwate Medical University School of Medicine, Morioka, Japan
| |
Collapse
|
71
|
Rauch A, Hennig D, Schäfer C, Wirth M, Marx C, Heinzel T, Schneider G, Krämer OH. Survivin and YM155: how faithful is the liaison? Biochim Biophys Acta Rev Cancer 2014; 1845:202-20. [PMID: 24440709 DOI: 10.1016/j.bbcan.2014.01.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/01/2014] [Accepted: 01/04/2014] [Indexed: 02/07/2023]
Abstract
Survivin belongs to the family of apoptosis inhibitors (IAPs), which antagonizes the induction of cell death. Dysregulated expression of IAPs is frequently observed in cancers, and the high levels of survivin in tumors compared to normal adult tissues make it an attractive target for pharmacological interventions. The small imidazolium-based compound YM155 has recently been reported to block the expression of survivin via inhibition of the survivin promoter. Recent data, however, question that this is the sole and main effect of this drug, which is already being tested in ongoing clinical studies. Here, we critically review the current data on YM155 and other new experimental agents supposed to antagonize survivin. We summarize how cells from various tumor entities and with differential expression of the tumor suppressor p53 respond to this agent in vitro and as murine xenografts. Additionally, we recapitulate clinical trials conducted with YM155. Our article further considers the potency of YM155 in combination with other anti-cancer agents and epigenetic modulators. We also assess state-of-the-art data on the sometimes very promiscuous molecular mechanisms affected by YM155 in cancer cells.
Collapse
Affiliation(s)
- Anke Rauch
- Center for Molecular Biomedicine, Institute for Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University of Jena, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - Dorle Hennig
- Center for Molecular Biomedicine, Institute for Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University of Jena, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - Claudia Schäfer
- Center for Molecular Biomedicine, Institute for Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University of Jena, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - Matthias Wirth
- II Department of Internal Medicine, Technical University of Munich, Munich, Germany
| | - Christian Marx
- Center for Molecular Biomedicine, Institute for Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University of Jena, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - Thorsten Heinzel
- Center for Molecular Biomedicine, Institute for Biochemistry and Biophysics, Department of Biochemistry, Friedrich Schiller University of Jena, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - Günter Schneider
- II Department of Internal Medicine, Technical University of Munich, Munich, Germany
| | - Oliver H Krämer
- Department of Toxicology, University Medical Center, Obere Zahlbacher Str. 67, 55131 Mainz, Germany.
| |
Collapse
|
72
|
Garcia PB, Attardi LD. Illuminating p53 function in cancer with genetically engineered mouse models. Semin Cell Dev Biol 2014; 27:74-85. [PMID: 24394915 DOI: 10.1016/j.semcdb.2013.12.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/06/2013] [Accepted: 12/23/2013] [Indexed: 01/23/2023]
Abstract
The key role of the p53 protein in tumor suppression is highlighted by its frequent mutation in human cancers and by the completely penetrant cancer predisposition of p53 null mice. Beyond providing definitive evidence for the critical function of p53 in tumor suppression, genetically engineered mouse models have offered numerous additional insights into p53 function. p53 knock-in mice expressing tumor-derived p53 mutants have revealed that these mutants display gain-of-function activities that actively promote carcinogenesis. The generation of p53 knock-in mutants with alterations in different domains of p53 has helped further elucidate the cellular and biochemical activities of p53 that are most fundamental for tumor suppression. In addition, modulation of p53 post-translational modification (PTM) status by generating p53 knock-in mouse strains with mutations in p53 PTM sites has revealed a subtlety and complexity to p53 regulation. Analyses of mouse models perturbing upstream regulators of p53 have solidified the notion that the p53 pathway can be compromised by means other than direct p53 mutation. Finally, switchable p53 models that allow p53 reactivation in tumors have helped evaluate the potential of p53 restoration therapy for cancer treatment. Collectively, mouse models have greatly enhanced our understanding of physiological p53 function and will continue to provide new biological and clinical insights in future investigations.
Collapse
Affiliation(s)
- Patty B Garcia
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
73
|
Haque S, Yan XJ, Rosen L, McCormick S, Chiorazzi N, Mongini PKA. Effects of prostaglandin E2 on p53 mRNA transcription and p53 mutagenesis during T-cell-independent human B-cell clonal expansion. FASEB J 2013; 28:627-43. [PMID: 24145719 DOI: 10.1096/fj.13-237792] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Within T-cell-dependent germinal centers, p53 gene transcription is repressed by Bcl-6 and is thus less vulnerable to mutation. Malignant lymphomas within inflamed extranodal sites exhibit a relatively high incidence of p53 mutations. The latter might originate from normal B-cell clones manifesting activation-induced cytosine deaminase (AID) and up-regulated p53 following T-cell-independent (TI) stimulation. We here examine p53 gene transcription in such TI clones, with a focus on modulatory effects of prostaglandin E2 (PGE2), and evaluate progeny for p53 mutations. Resting IgM(+)IgD(+)CD27(-) B cells from human tonsils were labeled with CFSE and stimulated in vitro with complement-coated antigen surrogate, IL-4, and BAFF ± exogenous PGE2 (50 nM) or an analog specific for the EP2 PGE2 receptor. We use flow cytometry to measure p53 and AID protein within variably divided blasts, qRT-PCR of p53 mRNA from cultures with or without actinomycin D to monitor mRNA transcription/stability, and single-cell p53 RT-PCR/sequencing to assess progeny for p53 mutations. We report that EP2 signaling triggers increased p53 gene transcriptional activity in AID(+) cycling blasts (P<0.01). Progeny exhibit p53 mutations at a frequency (8.5 × 10(-4)) greater than the baseline error rate (<0.8 × 10(-4)). We conclude that, devoid of the repressive influences of Bcl-6, dividing B lymphoblasts in inflamed tissues should display heightened p53 transcription and increased risk of p53 mutagenesis.
Collapse
Affiliation(s)
- Shabirul Haque
- 1Laboratory of B-Cell Biology, Karches Center for CLL Research and Center for Autoimmunity and Musculoskeletal Diseases, Feinstein Institute for Medical Research, 350 Community Dr., Manhasset, NY 11030, USA.
| | | | | | | | | | | |
Collapse
|
74
|
Azzam G, Wang X, Bell D, Murphy ME. CSF1 is a novel p53 target gene whose protein product functions in a feed-forward manner to suppress apoptosis and enhance p53-mediated growth arrest. PLoS One 2013; 8:e74297. [PMID: 24019961 PMCID: PMC3760869 DOI: 10.1371/journal.pone.0074297] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 07/25/2013] [Indexed: 12/29/2022] Open
Abstract
The p53 tumor suppressor gene has a common polymorphism at codon 72 that alters its function. We previously reported that the proline 72 polymorphic variant of p53 (P72) demonstrates increased ability to transactivate a subset of genes, relative to arginine 72 (R72); one of these genes is macrophage colony stimulating factor (CSF1). At present, the mechanism(s) underlying the increased transcriptional activity of P72 toward genes like CSF1 have not been completely elucidated. Additionally, the consequences of increased transcription of genes like CSF1 by the P72 variant to the downstream p53 pathway are unknown. In this report, we address these issues. We show that the CSF1 gene contains a conserved binding site for p53, and interestingly that the P72 variant shows increased ability to bind to this site. Moreover, we show that increased CSF1/CSF1R signaling in P72 cells feeds back on the p53 pathway to enhance p53 phosphorylation, levels, and transactivation of target genes, particularly the cyclin-dependent kinase inhibitor p21 (CDKN1A). This leads to an increase in p53-mediated growth arrest, along with a concomitant decrease in apoptosis. Notably, the CSF1/CSF1R signaling axis is overexpressed in several epithelial cancers, and there is clinical evidence that this pathway plays a role in radio-resistance of some cancers. We show that cells expressing CSF1 and CSF1R are indeed radio-resistant, and further, that this effect requires p53. These combined data are the first to implicate the CSF1/CSF1R pathway in the decision between p53-mediated growth arrest and apoptosis. They are also the first to highlight a cytokine as influential in cell fate determined by p53 in epithelial cells. Finally, these data may explain the association of the P72 variant and the CSF1/CSF1R pathway with increased senescence and radio-resistance in some epithelial tumor types.
Collapse
Affiliation(s)
- Gregory Azzam
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Xuting Wang
- Laboratory of Molecular Genetics, Intramural Research Program, National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Douglas Bell
- Laboratory of Molecular Genetics, Intramural Research Program, National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Maureen E. Murphy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
75
|
Hoesel B, Schmid JA. The complexity of NF-κB signaling in inflammation and cancer. Mol Cancer 2013; 12:86. [PMID: 23915189 PMCID: PMC3750319 DOI: 10.1186/1476-4598-12-86] [Citation(s) in RCA: 2374] [Impact Index Per Article: 215.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/30/2013] [Indexed: 02/07/2023] Open
Abstract
The NF-κB family of transcription factors has an essential role in inflammation and innate immunity. Furthermore, NF-κB is increasingly recognized as a crucial player in many steps of cancer initiation and progression. During these latter processes NF-κB cooperates with multiple other signaling molecules and pathways. Prominent nodes of crosstalk are mediated by other transcription factors such as STAT3 and p53 or the ETS related gene ERG. These transcription factors either directly interact with NF-κB subunits or affect NF-κB target genes. Crosstalk can also occur through different kinases, such as GSK3-β, p38, or PI3K, which modulate NF-κB transcriptional activity or affect upstream signaling pathways. Other classes of molecules that act as nodes of crosstalk are reactive oxygen species and miRNAs. In this review, we provide an overview of the most relevant modes of crosstalk and cooperativity between NF-κB and other signaling molecules during inflammation and cancer.
Collapse
Affiliation(s)
- Bastian Hoesel
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Johannes A Schmid
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| |
Collapse
|
76
|
Abstract
PURPOSE OF REVIEW The p53 tumor suppressor is a master regulator of antitumor defenses through its control of growth arrest, senescence and apoptosis. In recent years, p53 regulation was found to extend to a variety of biological processes including autophagy, fertility, metabolism and immune responses. Here, we focus on the role of p53 in the immune system. We explore the relationship between p53 and the innate immune response with particular emphasis on the Toll-like receptor (TLR) pathway and implications for cancer therapy. RECENT FINDINGS Numerous studies have shown that the immune system, especially innate immunity, has a critical role in tumor development. It appears that p53 can influence innate immune responses as part of its tumor suppressor activities and recent work suggests that the complete set of innate immune TLR genes are responsive to chromosomal stress and the transcriptional network regulated by p53. Activation of p53 by common antitumor agents results in p53 dependent regulation of expression of most TLR genes in human primary and cancer cell lines, resulting in modulation of TLR downstream responses to cognate ligands. In addition several tumor-associated p53 mutants can also affect TLR gene expression. These observations together with the discovery of other immune-related p53 target genes provide new insights into the relationship between p53 and immunity and suggest approaches that might be useful in cancer therapies. SUMMARY The tumor suppressor p53 can modulate innate immune gene responses in response to factors that can activate p53. This is expected to provide new opportunities in cancer diagnosis and in chemotherapeutic strategies that employ specific TLR agonists or antagonists that target the TLR pathway.
Collapse
|
77
|
Liang M, Yao G, Yin M, Lü M, Tian H, Liu L, Lian J, Huang X, Sun F. Transcriptional cooperation between p53 and NF-κB p65 regulates microRNA-224 transcription in mouse ovarian granulosa cells. Mol Cell Endocrinol 2013; 370:119-29. [PMID: 23474441 DOI: 10.1016/j.mce.2013.02.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 12/24/2012] [Accepted: 02/19/2013] [Indexed: 10/27/2022]
Abstract
MicroRNAs (miRNAs) have been indicated to play key roles in ovarian follicular development. However, little is known about how the miRNA gene expression itself is regulated in the mammalian ovary. We previously reported that miR-224 is involved in TGF-β1-mediated follicular granulosa cell (GC) growth and estradiol (E2) production by targeting Smad4. Here, the transcriptional regulation of miR-224 expression in GCs was further investigated. Our results showed that both the tumor suppressor gene p53 and NF-κB p65 subunit suppressed the TGF-β1-induced increase in pri-miR-224 expression in GCs. ChIP assays demonstrated that TGF-β1 enhanced the binding of p53 and p65 to the proximal promoter region of GABAA receptor ε subunit (miR-224 host gene). p53 and p65 transcriptionally cooperated to inactivate the GABAA receptor ε subunit promoter. In addition, p53/p65 could up-regulate Smad4 expression by inhibiting its target miR-224 in GCs which contributed, at least partially, to the effects of miR-224 and Smad4 on GC proliferation and E2 release. Our results provide new data about the interplay between transcription factors involved in GC proliferation and function by cooperatively regulating miRNA expression.
Collapse
Affiliation(s)
- Meng Liang
- Department of Cell and Developmental Biology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Sarkar J, Dominguez E, Li G, Kusewitt DF, Johnson DG. Modeling gene-environment interactions in oral cavity and esophageal cancers demonstrates a role for the p53 R72P polymorphism in modulating susceptibility. Mol Carcinog 2013; 53:648-58. [PMID: 23475592 DOI: 10.1002/mc.22019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 01/24/2013] [Accepted: 02/04/2013] [Indexed: 12/14/2022]
Abstract
A large number of epidemiological studies have linked a common single-nucleotide polymorphism (SNP) in the human p53 gene to risk for developing a variety of cancers. This SNP encodes either an arginine or proline at position 72 (R72P) of the p53 protein, which can alter the apoptotic activity of p53 via transcriptional and non-transcriptional mechanisms. This SNP has also been reported to modulate the development of human papilloma virus (HPV)-driven cancers through differential targeting of the p53 variant proteins by the E6 viral oncoprotein. Mouse models for the p53 R72P polymorphism have recently been developed but a role for this SNP in modifying cancer risk in response to viral and chemical carcinogens has yet to be established experimentally. Here, we demonstrate that the p53 R72P polymorphism modulates the hyperprolferative, apoptotic and inflammatory phenotypes caused by expression of the HPV16 E6 and E7 oncoproteins. Moreover, the R72P SNP also modifies the carcinogenic response to the chemical carcinogen 4NQO, in the presence and absence of the HPV16 transgene. Our findings confirm several human epidemiological studies associating the codon 72 proline variant with increased risk for certain cancers but also suggest that there are tissue-specific differences in how the R72P polymorphism influences the response to environmental carcinogens.
Collapse
Affiliation(s)
- Jayanta Sarkar
- Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, Texas
| | | | | | | | | |
Collapse
|
79
|
The p53 Codon 72 Polymorphism (rs1042522) Is Associated with Proliferative Vitreoretinopathy. Ophthalmology 2013. [DOI: 10.1016/j.ophtha.2012.08.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
80
|
Patel KR, Vajaria BN, Begum R, Shah FD, Patel JB, Shukla SN, Patel PS. Association between p53 Gene Variants and Oral Cancer Susceptibility in Population from Gujarat, West India. Asian Pac J Cancer Prev 2013; 14:1093-100. [DOI: 10.7314/apjcp.2013.14.2.1093] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
81
|
Leu JIJ, Murphy ME, George DL. The p53 Codon 72 Polymorphism Modifies the Cellular Response to Inflammatory Challenge in the Liver. ACTA ACUST UNITED AC 2013; 2. [PMID: 23991369 DOI: 10.4172/2167-0889.1000117] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The p53 protein is a critical stress-response mediator and signal coordinator in cellular metabolism and environmental exposure to deleterious agents. In human populations, the p53 gene contains a common single nucleotide polymorphism (SNP) affecting codon 72 that determines whether a proline (P72) or an arginine (R72) is present at this amino acid position of the polypeptide. Previous studies carried out using human populations, mouse models, and cell culture analyses have provided evidence that this amino acid difference can alter p53 functional activities, and potentially also can affect clinical presentation of disease. The clinical presentation associated with many forms of liver disease is variable, but few of the responsible underlying genetic factors or molecular pathways have been identified. The aim of the present study was to investigate whether the p53 codon 72 polymorphism influences the cellular response to hepatic stresses. A humanized p53 knock-in (Hupki) mouse model was used to address this issue. Mice expressing either the P72 or R72 normal variation of p53 were given an acute-, intermittent- or a chronic challenge, associated with exposure to lipopolysaccharide, D-galactosamine, or a high-fat diet. The results reveal that the livers of the P72 and R72 mice exhibit notable differences in inflammatory and apoptotic response to these distinct forms of stress. Interestingly the influence of this polymorphism on the response to stress is context dependent, with P72 showing increased response to liver toxins (lipopolysaccharide and D-galactosamine), but R72 showing increased response to metabolic stress (high fat diet). When taken together, these data point to the p53 codon 72 polymorphism as an important molecular mediator of events contributing to hepatic inflammation and metabolic homeostasis.
Collapse
Affiliation(s)
- Julia I-Ju Leu
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | |
Collapse
|
82
|
Interactions of miR-34b/c and TP53 polymorphisms on the risk of intracranial aneurysm. Clin Dev Immunol 2012; 2012:567586. [PMID: 22844323 PMCID: PMC3403301 DOI: 10.1155/2012/567586] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 05/22/2012] [Indexed: 02/06/2023]
Abstract
Several lines of evidence indicate that inflammatory processes play a key role in the happening and development of intracranial aneurysm (IA). Recently, polymorphisms in the TP53 gene were shown to be associated with inflammation and inflammatory disease. The aim of this study was to investigate the interactions of miR-34b/c and TP53 Arg72-Pro polymorphisms on the risk of IA in a Chinese population. A total of 590 individuals (including 164 patients with IA and 426 controls) were involved in this study. The polymorphisms (i.e., miR-34b/c rs4938723 and TP53 Arg72-Pro) were genotyped by polymerase chain reaction-restriction fragment length polymorphism assay and DNA sequencing. We found that the CC genotype of miR-34b/c rs4938723 was significantly associated with a decreased risk of IA compared with the TT genotype. Moreover, a significant gene interaction of the carriers with the combined genotypes of miR-34b/c rs4938723CC and TP53 Arg72Pro CG/CC/GG had a decreased risk of IA, compared with those carrying miR-34b/c rs4938723CT/TT+TP53 Arg72Pro GG/CG/CC combined genotypes. These findings suggest that the miR-34b/c rs4938723CC and TP53 Arg72-Pro polymorphisms may be involved in the susceptibility to IA.
Collapse
|
83
|
Sabatel H, Di Valentin E, Gloire G, Dequiedt F, Piette J, Habraken Y. Phosphorylation of p65(RelA) on Ser(547) by ATM represses NF-κB-dependent transcription of specific genes after genotoxic stress. PLoS One 2012; 7:e38246. [PMID: 22715377 PMCID: PMC3371017 DOI: 10.1371/journal.pone.0038246] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 05/02/2012] [Indexed: 12/03/2022] Open
Abstract
The NF-κB pathway is involved in immune and inflammation responses, proliferation, differentiation and cell death or survival. It is activated by many external stimuli including genotoxic stress. DNA double-strand breaks activate NF-κB in an ATM-dependent manner. In this manuscript, a direct interaction between p65(RelA) and the N-terminal extremity of ATM is reported. We also report that only one of the five potential ATM-(S/T)Q target sites present in p65, namely Ser547, is specifically phosphorylated by ATM in vitro. A comparative transcriptomic analysis performed in HEK-293 cells expressing either wild-type HA-p65 or a non-phosphorylatable mutant HA-p65S547A identified several differentially transcribed genes after an etoposide treatment (e.g. IL8, A20, SELE). The transcription of these genes is increased in cells expressing the mutant. Substitution of Ser547 to alanine does not affect p65 binding abilities on the κB site of the IL8 promoter but reduces p65 interaction with HDAC1. Cells expressing p65S547A have a higher level of histone H3 acetylated on Lys9 at the IL8 promoter, which is in agreement with the higher gene induction observed. These results indicate that ATM regulates a sub-set of NF-κB dependent genes after a genotoxic stress by direct phosphorylation of p65.
Collapse
Affiliation(s)
- Hélène Sabatel
- Laboratory of Virology and Immunology, GIGA-R, Signal Transduction Unit, University of Liège, Liège, Belgium
| | - Emmanuel Di Valentin
- Laboratory of Virology and Immunology, GIGA-R, Signal Transduction Unit, University of Liège, Liège, Belgium
| | - Geoffrey Gloire
- Laboratory of Virology and Immunology, GIGA-R, Signal Transduction Unit, University of Liège, Liège, Belgium
- Interface Entreprises-Université Liège Science Park, Angleur, Belgium
| | - Franck Dequiedt
- Laboratory of Signalisation and Protein Interaction, GIGA-R, Signal Transduction Unit, University of Liège, Liège, Belgium
| | - Jacques Piette
- Laboratory of Virology and Immunology, GIGA-R, Signal Transduction Unit, University of Liège, Liège, Belgium
| | - Yvette Habraken
- Laboratory of Virology and Immunology, GIGA-R, Signal Transduction Unit, University of Liège, Liège, Belgium
- * E-mail:
| |
Collapse
|
84
|
Loeb KR, Asgari MM, Hawes SE, Feng Q, Stern JE, Jiang M, Argenyi ZB, de Villiers EM, Kiviat NB. Analysis of Tp53 codon 72 polymorphisms, Tp53 mutations, and HPV infection in cutaneous squamous cell carcinomas. PLoS One 2012; 7:e34422. [PMID: 22545084 PMCID: PMC3335843 DOI: 10.1371/journal.pone.0034422] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 02/28/2012] [Indexed: 12/26/2022] Open
Abstract
Background Non-melanoma skin cancers are one of the most common human malignancies accounting for 2–3% of tumors in the US and represent a significant health burden. Epidemiology studies have implicated Tp53 mutations triggered by UV exposure, and human papilloma virus (HPV) infection to be significant causes of non-melanoma skin cancer. However, the relationship between Tp53 and cutaneous HPV infection is not well understood in skin cancers. In this study we assessed the association of HPV infection and Tp53 polymorphisms and mutations in lesional specimens with squamous cell carcinomas. Methods We studied 55 cases of histologically confirmed cutaneous squamous cell carcinoma and 41 controls for the presence of HPV infection and Tp53 genotype (mutations and polymorphism). Results We found an increased number of Tp53 mutations in the squamous cell carcinoma samples compared with perilesional or control samples. There was increased frequency of homozygous Tp53-72R polymorphism in cases with squamous cell carcinomas, while the Tp53-72P allele (Tp53-72R/P and Tp53-72P/P) was more frequent in normal control samples. Carcinoma samples positive for HPV showed a decreased frequency of Tp53 mutations compared to those without HPV infection. In addition, carcinoma samples with a Tp53-72P allele showed an increased incidence of Tp53 mutations in comparison carcinomas samples homozygous for Tp53-72R. Conclusions These studies suggest there are two separate pathways (HPV infection and Tp53 mutation) leading to cutaneous squamous cell carcinomas stratified by the Tp53 codon-72 polymorphism. The presence of a Tp53-72P allele is protective against cutaneous squamous cell carcinoma, and carcinoma specimens with Tp53-72P are more likely to have Tp53 mutations. In contrast Tp53-72R is a significant risk factor for cutaneous squamous cell carcinoma and is frequently associated with HPV infection instead of Tp53 mutations. Heterozygosity for Tp53-72R/P is protective against squamous cell carcinomas, possibly reflecting a requirement for both HPV infection and Tp53 mutations.
Collapse
Affiliation(s)
- Keith R. Loeb
- Divisions of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Maryam M. Asgari
- Department of Epidemiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Stephen E. Hawes
- Department of Epidemiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Qinghua Feng
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Joshua E. Stern
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Mingjun Jiang
- Institute of Dermatology, National Academy of Medical Sciences, Nanjing, People’s Republic of China
| | - Zsolt B. Argenyi
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Division of Dermatology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Ethel-Michele de Villiers
- Division for the Characterization of Tumorviruses, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Nancy B. Kiviat
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
85
|
Wasserman JD, Zambetti GP, Malkin D. Towards an understanding of the role of p53 in adrenocortical carcinogenesis. Mol Cell Endocrinol 2012; 351:101-10. [PMID: 21930187 PMCID: PMC3288384 DOI: 10.1016/j.mce.2011.09.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 08/31/2011] [Accepted: 09/05/2011] [Indexed: 12/17/2022]
Abstract
Adrenocortical carcinoma (ACC) is recognized to be a component tumor of the Li Fraumeni Syndrome (LFS), a familial cancer predisposition resulting from germline mutations in the p53 tumor-suppressor. p53 activity is tightly regulated by multiple post-translational mechanisms, disruption of which may lead to tumorigenesis. ACC is present in disproportionately high rates among p53-mutation carriers, suggesting tissue-specific manifestations of p53 deficiency. Additionally, p53-associated ACC demonstrates a strong predominance in infants and children. Several of the p53 alleles associated with pediatric ACC, however, retain significant wild-type activity and demonstrate incomplete penetrance, a finding distinct from other LFS-component tumors. In this review, we discuss the relationship between p53 and adrenocortical carcinogenesis, with specific focus on disease-specific alleles, tumorigenesis in the context of adrenal development and potential therapeutic approaches to p53-associated ACC.
Collapse
Affiliation(s)
- Jonathan D. Wasserman
- Division of Endocrinology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada M5G 1X8
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Gerard P. Zambetti
- Department of Biochemistry, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678
| | - David Malkin
- Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada M5G 1X8
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Hematology/Oncology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| |
Collapse
|
86
|
Cherdyntseva NV, Denisov EV, Litviakov NV, Maksimov VN, Malinovskaya EA, Babyshkina NN, Slonimskaya EM, Voevoda MI, Choinzonov EL. Crosstalk Between the FGFR2 and TP53 Genes in Breast Cancer: Data from an Association Study and Epistatic Interaction Analysis. DNA Cell Biol 2012; 31:306-16. [DOI: 10.1089/dna.2011.1351] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Nadezhda V. Cherdyntseva
- Department of Experimental Oncology, Cancer Research Institute, Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russian Federation
| | - Evgeny V. Denisov
- Department of Experimental Oncology, Cancer Research Institute, Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russian Federation
| | - Nicolay V. Litviakov
- Department of Experimental Oncology, Cancer Research Institute, Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russian Federation
| | - Vladimir N. Maksimov
- Laboratory of Molecular Genetic Study of Internal Diseases, Institute of Internal Medicine, Siberian Branch of Russian Academy of Medical Sciences, Novosibirsk, Russian Federation
| | - Elena A. Malinovskaya
- Department of Experimental Oncology, Cancer Research Institute, Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russian Federation
| | - Natalia N. Babyshkina
- Department of Experimental Oncology, Cancer Research Institute, Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russian Federation
| | - Elena M. Slonimskaya
- Department of General Oncology, Cancer Research Institute, Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russian Federation
- Department of Oncology, Siberian State Medical University, Tomsk, Russian Federation
| | - Mikhail I. Voevoda
- Laboratory of Molecular Genetic Study of Internal Diseases, Institute of Internal Medicine, Siberian Branch of Russian Academy of Medical Sciences, Novosibirsk, Russian Federation
| | - Evgeny L. Choinzonov
- Department of Oncology, Siberian State Medical University, Tomsk, Russian Federation
- Department of Head and Neck Oncology, Cancer Research Institute, Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russian Federation
| |
Collapse
|
87
|
Abstract
It is only recently that the full importance of nuclear factor-κB (NF-κB) signalling to cancer development has been understood. Although much attention has focused on the upstream pathways leading to NF-κB activation, it is now becoming clear that the inhibitor of NF-κB kinases (IKKs), which regulate NF-κB activation, have many independent functions in tissue homeostasis and normal immune function that could compromise the clinical utility of IKK inhibitors. Therefore, if the NF-κB pathway is to be properly exploited as a target for both anticancer and anti-inflammatory drugs, it is appropriate to reconsider the complex roles of the individual NF-κB subunits.
Collapse
Affiliation(s)
- Neil D Perkins
- Institute for Cell and Molecular Biosciences, Newcastle University, Medical School, Catherine Cookson Building, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK.
| |
Collapse
|
88
|
Gudkov AV, Gurova KV, Komarova EA. Inflammation and p53: A Tale of Two Stresses. Genes Cancer 2011; 2:503-16. [PMID: 21779518 DOI: 10.1177/1947601911409747] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Numerous observations indicate a strong link between chronic inflammation and cancer. This link is supported by substantial experimental evidence indicating mutual negative regulation of NF-κB, the major regulator of inflammation, and p53, the major tumor suppressor. This antagonistic relationship reflects the opposite principles of the physiological responses driven by these transcription factors, which act as sensors and mediators of intrinsic and extrinsic cell stresses, respectively. Constitutive activation of NF-κB, the underlying cause of chronic inflammation, is a common acquired characteristic of tumors. A variety of experimental methods have been used to demonstrate that constitutive activation of NF-κB reduces the tumor suppressor activity of p53, thereby creating permissive conditions for dominant oncogene-mediated transformation. Loss of p53 activity is also a characteristic of the majority of tumors and results in unleashed inflammatory responses due to loss of p53-mediated NF-κB suppression. On the other hand, in natural or pharmacological situations of enforced p53 activation, NF-κB activity, inflammation, and immune responses are reduced, resulting in different pathologies. It is likely that the chronic inflammation that is commonly acquired in various tissues of older mammals leads to general suppression of p53 function, which would explain the increased risk of cancer observed in aging animals and humans. Although the molecular mechanisms underlying reciprocal negative regulation of p53 and NF-κB remain to be deciphered, this phenomenon has important implications for pharmacological prevention of cancer and aging and for new approaches to control inflammation.
Collapse
Affiliation(s)
- Andrei V Gudkov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | | |
Collapse
|
89
|
Sen T, Sen N, Huang Y, Sinha D, Luo ZG, Ratovitski EA, Sidransky D. Tumor protein p63/nuclear factor κB feedback loop in regulation of cell death. J Biol Chem 2011; 286:43204-13. [PMID: 22020940 DOI: 10.1074/jbc.m111.257105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor protein (TP)-p53 family members often play proapoptotic roles, whereas nuclear factor κB (NF-κB) functions as a proapoptotic and antiapoptotic regulator depending on the cellular environment. We previously showed that the NF-κB activation leads to the reduction of the TP63 isoform, ΔNp63α, thereby rendering the cells susceptible to cell death upon DNA damage. However, the functional relationship between TP63 isotypes and NF-κB is poorly understood. Here, we report that the TAp63 regulates NF-κB transcription and protein stability subsequently leading to the cell death phenotype. We found that TAp63α induced the expression of the p65 subunit of NF-κB (RELA) and target genes involved in cell cycle arrest or apoptosis, thereby triggering cell death pathways in MCF10A cells. RELA was shown to concomitantly modulate specific cell survival pathways, making it indispensable for the TAp63α-dependent regulation of cell death. We showed that TAp63α and RELA formed protein complexes resulted in their mutual stabilization and inhibition of the RELA ubiquitination. Finally, we showed that TAp63α directly induced RelA transcription by binding to and activating of its promoter and, in turn, leading to activation of the NF-κB-dependent cell death genes. Overall, our data defined the regulatory feedback loop between TAp63α and NF-κB involved in the activation of cell death process of cancer cells.
Collapse
Affiliation(s)
- Tanusree Sen
- Departments of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | | | | | | | | | | | | |
Collapse
|
90
|
Azzam GA, Frank AK, Hollstein M, Murphy ME. Tissue-specific apoptotic effects of the p53 codon 72 polymorphism in a mouse model. Cell Cycle 2011; 10:1352-5. [PMID: 21566457 DOI: 10.4161/cc.10.9.15344] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Currently there are several dozen human polymorphisms that have been loosely associated with cancer risk. Correlating such variants with cancer risk has been challenging, primarily due to factors such as genetic heterogeneity, contributions of diet and environmental factors, and the difficulty in obtaining large sample sizes for analysis. Such difficulties can be circumvented with the establishment of mouse models for human variants. Recently, several groups have modeled human cancer susceptibility polymorphisms in the mouse. Remarkably, in each case these mouse models have accurately reflected human phenotypes, and clarified the contribution of these variants to cancer risk. We recently reported on a mouse model for the codon 72 polymorphism in p53, and found that this polymorphism regulates the ability to cooperate with NF-kB and induce apoptosis. Here-in we present evidence that this polymorphism impacts the apoptotic function of p53 in a tissue-specific manner; such tissue-specific effects of polymorphic variants represent an added challenge to human cancer risk association studies. The data presented here support the premise that modeling human polymorphisms in the mouse represents a powerful tool to assess the impact of these variants on cancer risk, progression and therapy.
Collapse
|